Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 836
Filter
1.
Molecules ; 29(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38998978

ABSTRACT

The regulation of the cancer cell cycle heavily relies on cyclin-dependent kinases (CDKs). Targeting CDKs has been identified as a promising approach for effective cancer therapy. In recent years, there has been significant attention paid towards developing small-molecule CDK inhibitors in the field of drug discovery. Notably, five such inhibitors have already received regulatory approval for the treatment of different cancers, including breast tumors, lung malignancies, and hematological malignancies. This review provides an overview of the synthetic routes used to produce 17 representative small-molecule CDK inhibitors that have obtained regulatory approval or are currently being evaluated through clinical trials. It also discusses their clinical applications for treating CDK-related diseases and explores the challenges and limitations associated with their use in a clinical setting, which will stimulate the further development of novel CDK inhibitors. By integrating therapeutic applications, synthetic methodologies, and mechanisms of action observed in various clinical trials involving these CDK inhibitors, this review facilitates a comprehensive understanding of the versatile roles and therapeutic potential offered by interventions targeting CDKs.


Subject(s)
Antineoplastic Agents , Cyclin-Dependent Kinases , Neoplasms , Protein Kinase Inhibitors , Small Molecule Libraries , Humans , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Small Molecule Libraries/chemical synthesis , Animals , Drug Discovery , Clinical Trials as Topic
2.
Science ; 385(6704): 91-99, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38963839

ABSTRACT

Sickle cell disease (SCD) is a prevalent, life-threatening condition attributable to a heritable mutation in ß-hemoglobin. Therapeutic induction of fetal hemoglobin (HbF) can ameliorate disease complications and has been intently pursued. However, safe and effective small-molecule inducers of HbF remain elusive. We report the discovery of dWIZ-1 and dWIZ-2, molecular glue degraders of the WIZ transcription factor that robustly induce HbF in erythroblasts. Phenotypic screening of a cereblon (CRBN)-biased chemical library revealed WIZ as a previously unknown repressor of HbF. WIZ degradation is mediated by recruitment of WIZ(ZF7) to CRBN by dWIZ-1, as resolved by crystallography of the ternary complex. Pharmacological degradation of WIZ was well tolerated and induced HbF in humanized mice and cynomolgus monkeys. These findings establish WIZ degradation as a globally accessible therapeutic strategy for SCD.


Subject(s)
Anemia, Sickle Cell , Antisickling Agents , Fetal Hemoglobin , Kruppel-Like Transcription Factors , Nerve Tissue Proteins , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/metabolism , Antisickling Agents/chemistry , Antisickling Agents/pharmacology , Antisickling Agents/therapeutic use , Crystallography, X-Ray , Drug Discovery , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Kruppel-Like Transcription Factors/metabolism , Macaca fascicularis , Nerve Tissue Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
3.
Drug Discov Today ; 29(7): 104053, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849028

ABSTRACT

Pancreatic cancer (PC), a disease with high heterogeneity and a dense stromal microenvironment, presents significant challenges and a bleak prognosis. Recent breakthroughs have illuminated the crucial interplay among RAS, epidermal growth factor receptor (EGFR), and hedgehog pathways in PC progression. Small molecular inhibitors have emerged as a potential solution with their advantages of oral administration and the ability to target intracellular and extracellular sites effectively. However, despite the US FDA approving over 100 small-molecule targeted antitumor drugs, challenges such as low response rates and drug resistance persist. This review delves into the possibility of using small molecules to treat persistent or spreading PC, highlighting the challenges and the urgent need for a diverse selection of inhibitors to develop more effective treatment strategies.


Subject(s)
Antineoplastic Agents , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Animals , Drug Resistance, Neoplasm , Molecular Targeted Therapy , ErbB Receptors/antagonists & inhibitors , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
4.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892472

ABSTRACT

Triple-negative breast cancer (TNBC) cells are devoid of estrogen receptors (ERs), progesterone receptor (PRs), and human epidermal growth factor receptor 2 (HER2), and it (TNBC) counts for about 10-15% of all breast cancers. TNBC is highly invasive, having a faster growth rate and a higher risk of metastasis and recurrence. Still, chemotherapy is one of the widely used options for treating TNBC. This study reviewed the histological and molecular characterization of TNBC subtypes, signaling pathways that are aberrantly expressed, and small molecules targeting these pathways, as either single agents or in combination with other therapeutic agents like chemotherapeutics, immunotherapeutics, and antibody-drug conjugates; their mechanisms of action, challenges, and future perspectives were also reviewed. A detailed analytical review was carried out using the literature collected from the SciFinder, PubMed, ScienceDirect, Google Scholar, ACS, Springer, and Wiley databases. Several small molecule inhibitors were found to be therapeutics for treating TNBC. The mechanism of action and the different signaling pathways through which the small molecules exert their effects were studied, including clinical trials, if reported. These small molecule inhibitors include buparlisib, everolimus, vandetanib, apatinib, olaparib, salidroside, etc. Some of the signaling pathways involved in TNBC, including the VEGF, PARP, STAT3, MAPK, EGFR, P13K, and SRC pathways, were discussed. Due to the absence of these biomarkers, drug development for treating TNBC is challenging, with chemotherapy being the main therapeutic agent. However, chemotherapy is associated with chemoresistance and a high toxicity to healthy cells as side effects. Hence, there is a continuous demand for small-molecule inhibitors that specifically target several signaling pathways that are abnormally expressed in TNBC. We attempted to include all the recent developments in this field. Any omission is truly unintentional.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Female , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Signal Transduction/drug effects , Molecular Targeted Therapy , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
5.
Chemistry ; 30(38): e202401103, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38716707

ABSTRACT

This review covers the most recent advances in the development of inhibitors for the bacterial enzyme sortase A (SrtA). Sortase A (SrtA) is a critical virulence factor, present ubiquitously in Gram-positive bacteria of which many are pathogenic. Sortases are key enzymes regulating bacterial adherence to host cells, by anchoring extracellular matrix-binding proteins to the bacterial outer cell wall. By targeting virulence factors, effective treatment can be achieved, without inducing antibiotic resistance to the treatment. This is a potentially more sustainable, long-term approach to treating bacterial infections, including ones that display multiple resistance to current therapeutics. There are many promising approaches available for SrtA inhibition, some of which have the potential to advance into further clinical development, with peptidomimetic and in vivo active small molecules being among the most promising. There are currently no approved drugs on the market targeting SrtA, despite its promise, adding to the relevance of this review article, as it extends to the pharmaceutical industry additionally to academic researchers.


Subject(s)
Aminoacyltransferases , Anti-Bacterial Agents , Bacterial Proteins , Cysteine Endopeptidases , Peptidomimetics , Small Molecule Libraries , Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/chemistry , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Humans , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gram-Positive Bacteria/drug effects
6.
J Hematol Oncol ; 17(1): 30, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711100

ABSTRACT

As the most common form of epigenetic regulation by RNA, N6 methyladenosine (m6A) modification is closely involved in physiological processes, such as growth and development, stem cell renewal and differentiation, and DNA damage response. Meanwhile, its aberrant expression in cancer tissues promotes the development of malignant tumors, as well as plays important roles in proliferation, metastasis, drug resistance, immunity and prognosis. This close association between m6A and cancers has garnered substantial attention in recent years. An increasing number of small molecules have emerged as potential agents to target m6A regulators for cancer treatment. These molecules target the epigenetic level, enabling precise intervention in RNA modifications and efficiently disrupting the survival mechanisms of tumor cells, thus paving the way for novel approaches in cancer treatment. However, there is currently a lack of a comprehensive review on small molecules targeting m6A regulators for anti-tumor. Here, we have comprehensively summarized the classification and functions of m6A regulators, elucidating their interactions with the proliferation, metastasis, drug resistance, and immune responses in common cancers. Furthermore, we have provided a comprehensive overview on the development, mode of action, pharmacology and structure-activity relationships of small molecules targeting m6A regulators. Our aim is to offer insights for subsequent drug design and optimization, while also providing an outlook on future prospects for small molecule development targeting m6A.


Subject(s)
Adenosine , Antineoplastic Agents , Neoplasms , Small Molecule Libraries , Animals , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
7.
Molecules ; 29(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38731601

ABSTRACT

Alterations in cellular metabolism, such as dysregulation in glycolysis, lipid metabolism, and glutaminolysis in response to hypoxic and low-nutrient conditions within the tumor microenvironment, are well-recognized hallmarks of cancer. Therefore, understanding the interplay between aerobic glycolysis, lipid metabolism, and glutaminolysis is crucial for developing effective metabolism-based therapies for cancer, particularly in the context of colorectal cancer (CRC). In this regard, the present review explores the complex field of metabolic reprogramming in tumorigenesis and progression, providing insights into the current landscape of small molecule inhibitors targeting tumorigenic metabolic pathways and their implications for CRC treatment.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Tumor Microenvironment/drug effects , Animals , Glycolysis/drug effects , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Lipid Metabolism/drug effects , Metabolic Networks and Pathways/drug effects
8.
Pharmacol Ther ; 249: 108485, 2023 09.
Article in English | MEDLINE | ID: mdl-37406740

ABSTRACT

Autophagy is a conserved process in which the cytosolic materials are degraded and eventually recycled for cellular metabolism to maintain homeostasis. The dichotomous role of autophagy in pathogenesis is complicated. Accumulating reports have suggested that cytoprotective autophagy is responsible for tumor growth and progression. Autophagy inhibitors, such as chloroquine (CQ) and hydroxychloroquine (HCQ), are promising for treating malignancies or overcoming drug resistance in chemotherapy. With the rapid development of nanotechnology, nanomaterials also show autophagy-inhibitory effects or are reported as the carriers delivering autophagy inhibitors. In this review, we summarize the small-molecule compounds and nanomaterials inhibiting autophagic flux as well as the mechanisms involved. The nanocarrier-based drug delivery systems for autophagy inhibitors and their distinct advantages are also described. The progress of autophagy inhibitors for clinical applications is finally introduced, and their future perspectives are discussed.


Subject(s)
Autophagy , Nanostructures , Neoplasms , Small Molecule Libraries , Autophagy/drug effects , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Nanostructures/therapeutic use , Nanoparticle Drug Delivery System , Clinical Trials as Topic , Humans
9.
Int J Mol Sci ; 24(9)2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37175788

ABSTRACT

Over the past three years, significant progress has been made in the development of novel promising drug candidates against COVID-19. However, SARS-CoV-2 mutations resulting in the emergence of new viral strains that can be resistant to the drugs used currently in the clinic necessitate the development of novel potent and broad therapeutic agents targeting different vulnerable spots of the viral proteins. In this study, two deep learning generative models were developed and used in combination with molecular modeling tools for de novo design of small molecule compounds that can inhibit the catalytic activity of SARS-CoV-2 main protease (Mpro), an enzyme critically important for mediating viral replication and transcription. As a result, the seven best scoring compounds that exhibited low values of binding free energy comparable with those calculated for two potent inhibitors of Mpro, via the same computational protocol, were selected as the most probable inhibitors of the enzyme catalytic site. In light of the data obtained, the identified compounds are assumed to present promising scaffolds for the development of new potent and broad-spectrum drugs inhibiting SARS-CoV-2 Mpro, an attractive therapeutic target for anti-COVID-19 agents.


Subject(s)
Artificial Intelligence , COVID-19 Drug Treatment , Coronavirus 3C Proteases , Drug Discovery , Small Molecule Libraries , Models, Molecular , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Coronavirus 3C Proteases/antagonists & inhibitors , Drug Discovery/methods , Neural Networks, Computer
10.
Int J Mol Sci ; 23(11)2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35682885

ABSTRACT

Molecular glue (MG) compounds are a type of unique small molecule that can change the protein-protein interactions (PPIs) and interactomes by degrading, stabilizing, or activating the target protein after their binging. These small-molecule MGs are gradually being recognized for their potential application in treating human diseases, including cancer. Evidence suggests that small-molecule MG compounds could essentially target any proteins, which play critical roles in human disease etiology, where many of these protein targets were previously considered undruggable. Intriguingly, most MG compounds with high efficacy for cancer treatment can glue on and control multiple key protein targets. On the other hand, a single key protein target can also be glued by multiple MG compounds with distinct chemical structures. The high flexibility of MG-protein interaction profiles provides rich soil for the growth and development of small-molecule MG compounds that can be used as molecular tools to assist in unraveling disease mechanisms, and they can also facilitate drug development for the treatment of human disease, especially human cancer. In this review, we elucidate this concept by using various types of small-molecule MG compounds and their corresponding protein targets that have been documented in the literature.


Subject(s)
Neoplasms , Neurodegenerative Diseases , Humans , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Protein Binding , Proteins/metabolism , Proteolysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Bioorg Med Chem Lett ; 61: 128601, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35123003

ABSTRACT

Therapeutic approaches to stimulate regeneration and repair have the potential to transform healthcare and improve outcomes for patients suffering from numerous chronic degenerative diseases. To date most approaches have involved the transplantation of therapeutic cells, and while there have been a small number of clinical approvals, major hurdles exist to the routine adoption of such therapies. In recent years humans and other mammals have been shown to possess a regenerative capacity across multiple tissues and organs, and an innate regenerative and repair response has been shown to be activated in these organs in response to injury. These realisations have inspired a transformative approach in regenerative medicine: the development of new agents to directly target these innate regeneration and repair pathways. In this article we will review the current state of the art in the discovery of small molecule modulators of regeneration and their translation towards therapeutic agents, focussing specifically on the areas of neuroregeneration and cardiac regeneration.


Subject(s)
Neurodegenerative Diseases/drug therapy , Regenerative Medicine , Small Molecule Libraries/therapeutic use , Animals , Chronic Disease , Humans , Small Molecule Libraries/chemistry
13.
Nat Rev Drug Discov ; 21(1): 60-78, 2022 01.
Article in English | MEDLINE | ID: mdl-34535788

ABSTRACT

Integrins are cell adhesion and signalling proteins crucial to a wide range of biological functions. Effective marketed treatments have successfully targeted integrins αIIbß3, α4ß7/α4ß1 and αLß2 for cardiovascular diseases, inflammatory bowel disease/multiple sclerosis and dry eye disease, respectively. Yet, clinical development of others, notably within the RGD-binding subfamily of αv integrins, including αvß3, have faced significant challenges in the fields of cancer, ophthalmology and osteoporosis. New inhibitors of the related integrins αvß6 and αvß1 have recently come to the fore and are being investigated clinically for the treatment of fibrotic diseases, including idiopathic pulmonary fibrosis and nonalcoholic steatohepatitis. The design of integrin drugs may now be at a turning point, with opportunities to learn from previous clinical trials, to explore new modalities and to incorporate new findings in pharmacological and structural biology. This Review intertwines research from biological, clinical and medicinal chemistry disciplines to discuss historical and current RGD-binding integrin drug discovery, with an emphasis on small-molecule inhibitors of the αv integrins.


Subject(s)
Integrins/antagonists & inhibitors , Integrins/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Animals , Drug Discovery/methods , Humans , Protein Binding/drug effects
14.
J Med Chem ; 65(2): 1047-1131, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34624192

ABSTRACT

The central role of dysregulated kinase activity in the etiology of progressive disorders, including cancer, has fostered incremental efforts on drug discovery programs over the past 40 years. As a result, kinase inhibitors are today one of the most important classes of drugs. The FDA approved 73 small molecule kinase inhibitor drugs until September 2021, and additional inhibitors were approved by other regulatory agencies during that time. To complement the published literature on clinical kinase inhibitors, we have prepared a review that recaps this large data set into an accessible format for the medicinal chemistry community. Along with the therapeutic and pharmacological properties of each kinase inhibitor approved across the world until 2020, we provide the synthesis routes originally used during the discovery phase, many of which were only available in patent applications. In the last section, we also provide an update on kinase inhibitor drugs approved in 2021.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Approval , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Small Molecule Libraries/therapeutic use , Antineoplastic Agents/chemical synthesis , Humans , Neoplasms/enzymology , Protein Kinase Inhibitors/chemical synthesis , Small Molecule Libraries/chemical synthesis , United States , United States Food and Drug Administration
15.
Leukemia ; 36(3): 637-647, 2022 03.
Article in English | MEDLINE | ID: mdl-34711926

ABSTRACT

Aberrant RHO guanine nucleotide exchange factor (RhoGEF) activation is chief mechanism driving abnormal activation of their GTPase targets in transformation and tumorigenesis. Consequently, a small-molecule inhibitor of RhoGEF can make an anti-cancer drug. We used cellular, mouse, and humanized models of RAC-dependent BCR-ABL1-driven and Ph-like acute lymphoblastic leukemia to identify VAV3, a tyrosine phosphorylation-dependent RacGEF, as the target of the small molecule IODVA1. We show that through binding to VAV3, IODVA1 inhibits RAC activation and signaling and increases pro-apoptotic activity in BCR-ABL1-transformed cells. Consistent with this mechanism of action, cellular and animal models of BCR-ABL1-induced leukemia in Vav3-null background do not respond to IODVA1. By durably decreasing in vivo RAC signaling, IODVA1 eradicates leukemic propagating activity of TKI-resistant BCR-ABL1(T315I) B-ALL cells after treatment withdrawal. Importantly, IODVA1 suppresses the leukemic burden in the treatment refractory pediatric Ph+ and TKI-resistant Ph+ B-ALL patient-derived xenograft models better than standard-of-care dasatinib or ponatinib and provides a more durable response after treatment withdrawal. Pediatric leukemia samples with diverse genetic lesions show high sensitivity to IODVA1 ex vivo and this sensitivity is VAV3 dependent. IODVA1 thus spearheads a novel class of drugs that inhibits a RacGEF and holds promise as an anti-tumor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Female , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-vav/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Tumor Cells, Cultured
16.
Chem Biol Drug Des ; 99(1): 1-31, 2022 01.
Article in English | MEDLINE | ID: mdl-34148284

ABSTRACT

G-Quadruplex, a unique secondary structure in nucleic acids found throughout human genome, elicited widespread interest in the field of therapeutic research. Being present in key regulatory regions of oncogenes, RNAs and telomere, G-Quadruplex structure regulates transcription, translation, splicing, etc. Changes in its structure and stability leads to differential expression of oncogenes causing cancer. Thus, targeting G-Quadruplex structures with small molecules/other biologics has shown elevated research interest. Covering previous reports, in this review, we try to enlighten the facts on the structural diversity in G-Quadruplex ligands aiming to provide newer insights to design first-in-class drugs for the next-generation cancer treatment.


Subject(s)
Aptamers, Nucleotide/chemistry , G-Quadruplexes , Neoplasms/drug therapy , Peptides/chemistry , Small Molecule Libraries/chemistry , Aptamers, Nucleotide/therapeutic use , Drug Design , Humans , Ligands , Nucleic Acid Conformation , Peptides/therapeutic use , Small Molecule Libraries/therapeutic use
17.
Future Med Chem ; 14(4): 221-231, 2022 02.
Article in English | MEDLINE | ID: mdl-34939426

ABSTRACT

Background: Vitiligo is a relatively common depigmenting skin disorder. UV light stimulation is often used to obtain repigmentation. Wnt signaling regulates melanocyte differentiation, and expression of TYR is upregulated in narrow-band UVB-treated epidermis. Manipulation of these two pathways by drugs could serve as one of the therapeutic approaches for durable repigmentation. Methods & results: CD9 was identified as a novel TYR activator by virtual screening and bioactivity assay. CD9 activated the Wnt signaling pathway through triggering translocation of ß-catenin from cytoplasm to nucleus. Conclusion: The pathogenesis of vitiligo is complicated and varies with each individual, so combination therapy may be much more suitable for treatment of vitiligo. CD9 could synergize with other anti-inflammatory compounds or autoimmune suppressors to shorten repigmentation time and improve efficacy.


Subject(s)
Melanins/metabolism , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Animals , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Humans , Ligands , Mice , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Tetraspanin 29/chemistry , Tetraspanin 29/metabolism , Vitiligo/drug therapy , Wnt Signaling Pathway/drug effects
18.
Int J Mol Sci ; 22(24)2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34947997

ABSTRACT

Trace amine-associated receptor 1 (TAAR1) has emerged as a promising therapeutic target for neuropsychiatric disorders due to its ability to modulate monoaminergic and glutamatergic neurotransmission. In particular, agonist compounds have generated interest as potential treatments for schizophrenia and other psychoses due to TAAR1-mediated regulation of dopaminergic tone. Here, we review unmet needs in schizophrenia, the current state of knowledge in TAAR1 circuit biology and neuropharmacology, including preclinical behavioral, imaging, and cellular evidence in glutamatergic, dopaminergic and genetic models linked to the pathophysiology of psychotic, negative and cognitive symptoms. Clinical trial data for TAAR1 drug candidates are reviewed and contrasted with antipsychotics. The identification of endogenous TAAR1 ligands and subsequent development of small-molecule agonists has revealed antipsychotic-, anxiolytic-, and antidepressant-like properties, as well as pro-cognitive and REM-sleep suppressing effects of TAAR1 activation in rodents and non-human primates. Ulotaront, the first TAAR1 agonist to progress to randomized controlled clinical trials, has demonstrated efficacy in the treatment of schizophrenia, while another, ralmitaront, is currently being evaluated in clinical trials in schizophrenia. Coupled with the preclinical findings, this provides a rationale for further investigation and development of this new pharmacological class for the treatment of schizophrenia and other psychiatric disorders.


Subject(s)
Antipsychotic Agents/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Schizophrenia/drug therapy , Small Molecule Libraries/therapeutic use , Animals , Antipsychotic Agents/pharmacology , Clinical Trials as Topic , Disease Models, Animal , Dopamine/metabolism , Glutamic Acid/metabolism , Humans , Receptors, G-Protein-Coupled/agonists , Schizophrenia/metabolism , Small Molecule Libraries/pharmacology
19.
Biomolecules ; 11(12)2021 11 26.
Article in English | MEDLINE | ID: mdl-34944420

ABSTRACT

Lysosomal Storage Diseases are multisystemic disorders determined by genetic variants, which affect the proteins involved in lysosomal function and cellular metabolism. Different therapeutic approaches, which are based on the physiologic mechanisms that regulate lysosomal function, have been proposed for these diseases. Currently, enzyme replacement therapy, gene therapy, or small molecules have been approved or are under clinical development to treat lysosomal storage disorders. The present article reviews the main therapeutic strategies that have been proposed so far, highlighting possible limitations and future perspectives.


Subject(s)
Biomarkers/metabolism , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Clinical Trials as Topic , Enzyme Replacement Therapy , Gene Expression Regulation , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Humans , Lysosomal Storage Diseases/metabolism , Small Molecule Libraries/therapeutic use
20.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34830024

ABSTRACT

Drug resistance continues to be a major problem associated with cancer treatment. One of the primary causes of anticancer drug resistance is the frequently mutated RAS gene. In particular, considerable efforts have been made to treat KRAS-induced cancers by directly and indirectly controlling the activity of KRAS. However, the RAS protein is still one of the most prominent targets for drugs in cancer treatment. Recently, novel targeted protein degradation (TPD) strategies, such as proteolysis-targeting chimeras, have been developed to render "undruggable" targets druggable and overcome drug resistance and mutation problems. In this study, we discuss small-molecule inhibitors, TPD-based small-molecule chemicals for targeting RAS pathway proteins, and their potential applications for treating KRAS-mutant cancers. Novel TPD strategies are expected to serve as promising therapeutic methods for treating tumor patients with KRAS mutations.


Subject(s)
Neoplasms/drug therapy , Proteolysis/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , Small Molecule Libraries/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Mutation/drug effects , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...